Language selection

Search

Patent 2424394 Summary

Third-party information liability

Some of the information on this Web page has been provided by external sources. The Government of Canada is not responsible for the accuracy, reliability or currency of the information supplied by external sources. Users wishing to rely upon this information should consult directly with the source of the information. Content provided by external sources is not subject to official languages, privacy and accessibility requirements.

Claims and Abstract availability

Any discrepancies in the text and image of the Claims and Abstract are due to differing posting times. Text of the Claims and Abstract are posted:

  • At the time the application is open to public inspection;
  • At the time of issue of the patent (grant).
(12) Patent: (11) CA 2424394
(54) English Title: GROWTH FACTOR COMPLEX COMPRISING AN INSULIN-LIKE GROWTH FACTOR BINDING PROTEIN BOUND TO VITRONECTIN
(54) French Title: COMPLEXE DE FACTEURS DE CROISSANCE COMPRENANT UNE PROTEINE DE LIAISON DU FACTEUR DE CROISSANCE INSULINOIDE LIEE A LA VITRONECTINE
Status: Expired and beyond the Period of Reversal
Bibliographic Data
(51) International Patent Classification (IPC):
  • A61K 38/39 (2006.01)
  • A61K 38/27 (2006.01)
  • A61K 38/30 (2006.01)
  • A61K 38/31 (2006.01)
  • A61P 01/00 (2006.01)
  • A61P 05/00 (2006.01)
  • A61P 09/10 (2006.01)
  • A61P 19/04 (2006.01)
  • A61P 19/10 (2006.01)
  • A61P 37/00 (2006.01)
(72) Inventors :
  • UPTON, ZEE (Australia)
  • KRICKER, JENNIFER ANN (Australia)
(73) Owners :
  • FACTOR THERAPEUTICS LIMITED
(71) Applicants :
  • FACTOR THERAPEUTICS LIMITED (Australia)
(74) Agent: DEETH WILLIAMS WALL LLP
(74) Associate agent:
(45) Issued: 2017-05-23
(86) PCT Filing Date: 2001-09-24
(87) Open to Public Inspection: 2002-03-28
Examination requested: 2006-09-13
Availability of licence: N/A
Dedicated to the Public: N/A
(25) Language of filing: English

Patent Cooperation Treaty (PCT): Yes
(86) PCT Filing Number: PCT/AU2001/001196
(87) International Publication Number: AU2001001196
(85) National Entry: 2003-03-21

(30) Application Priority Data:
Application No. Country/Territory Date
PR 0309 (Australia) 2000-09-22

Abstracts

English Abstract


An isolated protein complex is provided which includes a growth factor, growth
factor binding protein and vitronectin. Preferably, the isolated protein
complex includes an insulin-like growth factor-I, insulin-like growth factor
binding protein-3 or insulin-like growth factor binding protein-5 and
vitronectin. Also provided are methods of modulating cell proliferation and/or
migration by administering said protein complex for the purposes of wound
healing, skin repair and tissue replacement therapy. Conversely, by using
agents that disrupt growth factor protein complexes formed in vivo, growth
factor-driven cell proliferation and/or migration may be suppressed such as
for the purposes of treating cancers, psoriasis, atherosclerosis and wounds
prone to hypertrophic scarring.


French Abstract

La présente invention concerne un complexe protéique isolé qui comprend un facteur de croissance, une protéine de liaison du facteur de croissance et de la vitronectine. De préférence, le complexe protéique isolé contient un facteur de croissance 1 de type insuline, une protéine 3 de liaison du facteur de croissance de type insuline ou une protéine 5 de liaison du facteur de croissance de type insuline et de la vitronectine. Cette invention concerne également des procédés qui permettent de moduler la prolifération cellulaire et/ou la migration des cellules au moyen de l'administration dudit complexe protéique pour guérir des blessures et pour la thérapie de réparation cutanée et de remplacement tissulaire. Inversement, l'utilisation d'agents qui altèrent les complexes protéiques de facteur de croissance formés in vivo permet de supprimer la prolifération et/ou la migration cellulaire pour traiter des cancers, le psoriasis, l'athérosclérose et des blessures prédisposées à la cicatrisation hypertrophique.

Claims

Note: Claims are shown in the official language in which they were submitted.


38
CLAIMS
1. An isolated protein complex comprising: insulin-like growth factor I
(IGF-I); an insulin-
like growth factor binding protein (IGFBP) selected from: IGFBP2, IGFBP3,
IGFBP4 and
IGFBP5; and vitronectin (VN).
2. The isolated protein complex of Claim 1, which is produced in
recombinant form.
3. The isolated protein complex of Claim 1, wherein the insulin-like growth
factor binding
protein is IGFBP3 or IGFBP5.
4. The isolated protein complex of any one of Claims 1-3, wherein the IGFBP
mediates
binding of IGF-I to VN.
5. An isolated protein complex comprising IGF-1, vitronectin and a non-
glycosylated
variant of an insulin-like growth factor binding protein (IGFBP) selected
from: IGFBP3,
IGFBP4 and IGFBP5.
6. An isolated protein complex comprising vitronectin and a variant growth
factor selected
from: des(1-3)IGF-I, des(1-6)IGF-II and insulin-like growth factor (IGF)-I
engineered to include
a heparin binding domain (HBD) comprising the amino acid sequence BXBBB,
wherein B is a
basic amino acid residue and X is any amino acid.
7. The isolated protein complex of Claim 6, wherein the variant growth
factor is IGF-I
engineered to include said HBD.
8. The isolated protein complex of Claim 6, consisting of: IGF-I engineered
to include said
HBD bound directly to vitronectin.
9. The isolated protein complex of Claim 6, wherein the HBD has the amino
acid sequence
KGRKR.
10. Use of a variant of an insulin-like growth factor binding protein
(IGFBP) selected from:
IGFBP2, IGFBP3, IGFBP4, and IGFBP5, said variant having a deleted or mutated
heparin
binding domain (HBD), said HBD normally comprising the amino acid sequence
BXBBB,
wherein B is a basic amino acid residue and X is any amino acid, to prevent or
disrupt formation
of the isolated protein complex of Claim 1.

39
11. Use according to Claim 10, wherein the variant comprises the mutated
amino acid
sequence MDGEA.
12. Use according to Claim 10 or Claim 11 wherein the variant IGFBP is
capable of binding
insulin-like growth factor I (IGF-I).
13. Use according to Claim 12 wherein the IGFBP variant is an antagonist.
14. The isolated protein complex of Claim 5, wherein the non-glycosylated
IGFBP is
IGFBP3.
15. The isolated protein complex of any one of Claims 1, 5, or 6, wherein
vitronectin is
multimeric.
16. The isolated protein complex of any one of Claims 1, 5, or 6, wherein
vitronectin is
monomeric.
17. A pharmaceutical composition comprising the isolated protein complex of
any one of
Claims 1, 5, or 6, and a pharmaceutically-acceptable carrier or diluent.
18. A surgical implant or prosthesis coated, impregnated or otherwise
pretreated with the
isolated protein complex of any one of Claims 1, 5, or 6.
19. A transformed cell engineered to express a recombinant protein complex
according to
any one of Claims 1, 5, or 6, or recombinant polypeptides capable of forming
said complex.
20. A method of modulating cell proliferation and/or migration including
the step of
administering to isolated animal epithelial cells or cells of epithelial
origin, an isolated protein
complex according to any one of Claims 1, 5, or 6.
21. A method of inhibiting or suppressing cell proliferation and/or
migration, including the
step of administering to isolated animal epithelial cells or cells of
epithelial origin, an agent
which prevents or disrupts formation of the isolated protein complex of any
one of Claims 1, 5,
or 6 wherein the agent prevents or disrupts an interaction between an IGFBP
and vitronectin,
wherein said agent is IGF-II or a variant IGF identified by a process
comprising testing whether
the agent prevents or disrupts formation of the isolated protein complex.
22. The method of Claim 21, wherein the agent is IGF-II or a variant of IGF-
II comprising
one of more amino acid sequence mutations.

40
23. Use of IGF-II or a variant of IGF-II comprising one of more amino acid
sequence
mutations to prevent or inhibit formation of the isolated protein complex of
Claim 1.
24. Use of an isolated protein complex according to any one of Claims 1, 5,
or 6 for the
manufacture of a medicament for modulating proliferation and/or migration of
epithelial cells or
cells of epithelial origin.
25. Use of an agent which prevents or disrupts formation of the isolated
protein complex of
any one of Claims 1, 5, or 6 for the manufacture of a medicament for
modulating proliferation
and/or migration of epithelial cells or cells of epithelial origin, wherein
the agent prevents or
disrupts an interaction between an IGFBP and vitronectin and wherein the agent
is identified by
a process comprising testing whether the agent prevents or disrupts
interaction between the
IGFBP and vitronectin, and wherein said agent is IGF-II or a variant IGF.
26. Use according to Claim 25, wherein the agent is IGF-II or a variant of
IGF-II comprising
one or more amino acid sequence mutations.
27. Use of the isolated protein complex of any one of Claims 1, 5, or 6,
for the preparation of
a medicament for the treatment of an epithelial cell disease characterized by
reduced
epithelialization.
28. Use according to Claim 27, wherein the epithelial cell disease causes
impaired gut
function through deterioration of the gastrointestinal epithelial lining.
29. Use of the isolated protein complex of any one of Claims 1, 5, or 6,
for promoting wound
healing, skin repair, ulcer healing, burn healing or in vitro skin
regeneration.
30. Use of the isolated protein complex of any one of Claims 1, 5, or 6,
for the preparation of
a medicament for promoting bone regeneration.
31. Use of the isolated protein complex of any one of Claims 1, 5, or 6,
for the preparation of
a medicament for promoting repair of damaged neuronal tissue.
32. Use of an agent that disrupts or prevents formation of the isolated
protein complex of any
one of Claims 1, 5, or 6, for the preparation of a medicament for treating
cancer, wherein the
agent prevents or disrupts an interaction between an IGFBP and vitronectin and
wherein the
agent is identified by a process comprising testing whether the agent prevents
or disrupts

41
interaction between the IGFBP and vitronectin, and wherein said agent is IGF-
II or a variant
IGF.
33. Use according to Claim 32, wherein the cancer is breast cancer.
34. Use of an agent that disrupts or prevents formation of the isolated
protein complex of any
one of Claims 1, 5, or 6, for the preparation of a medicament for treating
atherosclerosis,
wherein the agent prevents or disrupts an interaction between an IGFBP and
vitronectin, and
wherein said agent is IGF-II or a variant IGF.
35. Use of an agent that disrupts or prevents formation of the isolated
protein complex of any
one of Claims 1, 5, or 6, for the preparation of a medicament for treating
psoriasis, wherein the
agent prevents or disrupts an interaction between an IGFBP and vitronectin and
wherein the
agent is identified by a process comprising testing whether the agent prevents
or disrupts
interaction between the IGFBP and vitronectin, and wherein said agent is IGF-
II or a variant
IGF.
36. Use according to any one of Claims 32 to 35, wherein the agent is IGF-
II or a variant of
IGF-II comprising one or more amino acid sequence mutations.
37. Use of the isolated protein complex of any one of Claims 1, 5, or 6 in
the preparation of a
medicament for promoting an activity selected form the group consisting of
wound healing, skin
repair, ulcer healing and burn healing.

Description

Note: Descriptions are shown in the official language in which they were submitted.


CA 02424394 2010-05-31
1
GROWTH FACTOR COMPLEX COMPRISING AN INSULIN-LIKE GROWTH
FACTOR BINDING PROTEIN BOUND TO VITRONECTIN
FIELD OF THE INVENTION
THIS INVENTION relates to an isolated protein complex which includes a growth
factor
binding protein and vitronectin. In particular, this invention relates to an
isolated protein
complex which includes an insulin-like growth factor, an insulin-like growth
factor binding
protein and vitronectin. Also provided by the invention are growth factor
complexes comprising
variant growth factors and/or growth factor binding proteins that facilitate
or enhance formation
of the growth factor complexes. This invention also provides methods of
modulating cell
proliferation and/or migration by administering said protein complex for the
purposes of wound
healing, skin repair, cosmetic skin maintenance and tissue replacement
therapy. Conversely, by
disrupting protein complexes formed in vivo, growth factor-driven cell
proliferation and/or
migration can be suppressed such as for the purposes of treating cancers,
psoriasis,
atherosclerosis and wounds prone to hypertrophic scarring. These treatments
may have medical
and veterinary applications.
BACKGROUND OF THE INVENTION
Skin growth, repair and healing are subject to complex biological control
mechanisms
which act via both positive and negative signals. Such signals act at the
level of controlling cell
proliferation, differentiation and migration, and are typically mediated by
growth factor
polypeptides. In this regard, important growth factors include epidermal
growth factor (EGF)
and insulin-like growth factors (IGF- I and -II).
Human IGF-I has been reported to exert a wide range of biological activities
including
stimulation of cell proliferation, differentiation and migration, protection
from protein
degradation and apoptosis, as well as regulation of endocrine factors such as
growth hormone.
IGF-II has similar properties to IGF-I but appears to be more relevant to
carcinogenesis and fetal
and embryonic development, IGF-I having a greater role in postnatal
development.
Both IGF-I and IGF-II act through a binding interaction with the type I IGF
receptor
(IGFR). The availability of the IGFs for such an interaction is

CA 02424394 2003-03-21
WO 02/24219 PCT/AU01/01196
2
regulated by insulin like growth factor binding proteins (IGFBPs 1-6). IGFBPs
are
known to both positively and negatively regulate IGF function as well as
exhibit
IGF-independent activity.
Another functional component of IGF pathways is the type II
IGFR which is also known as the cation-independent mannose-6-phosphate
receptor (CI-MPR). The type II IGFR is a multifunctional protein that binds
lysosomal enzymes bearing marmose-6-phosphate moieties, as well as IGF-II,
although the functional significance of IGF-II binding is unclear (O'Dell &
Day,
1998, Int. J. Biochem. Cell Biol. 30 767; Braulke, 1999, Horm. Metab. Res. 31
242; Nykjaer et al., 1998, J. Cell. Biol. 141 815).
The IGFs have also been reported to bind another group of proteins
termed "IGFBP-related proteins" which share structural similarity and include
connective tissue growth factor (CTGF) and products encoded by the mac25, 120V
and cyr61 genes. These bind IGFs with much lower affinity than do IGFBPs.
More recently, vitronectin (VN) has been identified as an
extracellular matrix protein, structurally unrelated to IGFBPs and IGFBP-
related
proteins, that binds but
not IGF-I (Upton et al., 1999, Endocrinol. 140
2928).
Vitronectin is an ¨751(D, glycosylated extracellular matrix protein
which is also found in blood, and has been implicated in cancers, bone
diseases
and pathological disorders involving angiogenesis (reviewed in Schvartz et
al.,
1999, Int. J. Biochem. Cell Biol. 31 539). The role of vitronectin in events
such as
angiogenesis and tumorigenesis at least partly resides in the ability of
vitronectin
to bind integrins and to interact with components of the urokinase plasminogen
activator system (for example PM-1, uPAR, plasminogen) to thereby promote cell
proliferation, adhesion, spreading and migration. Vitronectin has more
specifically been implicated in preventing tumor cell apoptosis in response to
drug
treatment (Uhm et al., 1999, Clin. Cancer Res. 5 1587). Vitronectin appears to
be
a carrier of IGF-II in the circulation (McMurtry et al., 1996, J. Endocrinol.
150
149).
OBJECT OF THE INVENTION

CA 02424394 2003-03-21
PCIYAU01/01196
Received 21 August 2002
3
The present inventors have surprisingly discovered that IGFBPs
bind vitronectin, and that IGF-1 can bind vitronectin when bound to an IGFBP.
It is therefore an object of the invention to provide an isolated
IGFBP and vitronectin-containing complex.
SUMMARY OF THE INVENTION
In a first aspect, the invention provides an isolated polypeptide
complex comprising a growth factor binding protein and vitronectin.
Preferably, the isolated polypeptide complex further comprises a
growth factor.
A preferred growth factor is insulin-like growth factor-I (IGF-I).
Other growth factors include epidermal growth factor (EGF),
fibroblast growth factor (FGF), basic fibroblast growth factor (bFGF),
osteopontin, PAT-1 and transferrin.
Other biologically active proteins that may form protein complexes
of the invention include thrombospondin-1, tenascin-C, PM-1, plasminogen,
fibrinogen, and fibrin.
A preferred growth factor binding protein is an insulin-like growth
factor binding protein selected from the group consisting of IGFBP1, IGFBP2,
IGFBP3, IGFBP4, IGFBP5 and IGFBP6.
A more preferred insulin-like growth factor binding protein is
IGFBP2, IGFBP3, IGFBP4 or IGFBP5.
An even more preferred insulin-like growth factor binding protein
is IGFBP3 or IGFBP5.
In a second aspect, the invention provides an isolated protein
complex comprising an IGFBP-related protein and vitronectin.
Preferably, the isolated polypeptide complex further comprises a
growth factor, preferably IGF-I.
In one embodiment, the IGFBP-related protein is selected from the
group consisting of connective tissue growth factor (CTGF), a polypeptide
encoded by the mac25 gene, a polypeptide encoded by the nov gene and a
polypeptide encoded by the cyr61 gene.
In a third aspect, the invention provides a isolated protein complex
AMENDED SHEET
a5mAkm

CA 02424394 2003-03-21
PC T/AU01/01196
Received 21 August 2002
4
comprising vitronectin, a variant growth factor and/or a variant growth factor
binding protein.
In one embodiment, the isolated protein complex of this aspect
comprises vitronectin and a variant growth factor engineered to include a
heparin
binding domain (HBD).
In another embodiment, the isolated protein complex of this aspect
comprises vitronectin and a non-glycosylated growth factor binding protein.
In yet another embodiment, the isolated protein complex of this
aspect comprises vitronectin and a variant growth factor selected from the
group
consisting of des(1-6)IGF-I1 and des(1-3)IGF-I.
Also contemplated according to the first- second- and third-
mentioned aspects is that the isolated protein complex of the invention may
further include an acid-labile subunit, a polypeptide which can complex with
an
IGFBP, referred to hereinafter as ALS.
The invention according to the first-, second- and third-mentioned
aspects also contemplates isolated protein complexes comprising variants and
biologically-active fragments of growth factors, growth factor binding
proteins,
IGFBP-related proteins and vitronectin, and use of such complexes.
Biologically-
active fragments and variants include within their scope analogues, mutants,
agonists and antagonists of said growth factors, growth factor binding
proteins,
IGFBP-related proteins and vitronectin.
In a fourth aspect, the invention provides a pharmaceutical
composition comprising one or more isolated protein complexes according to the
first-, second- or third-mentioned aspect and a pharmaceutically-acceptable
carrier
or diluent.
In a fifth aspect, the invention provides a pharmaceutical
composition comprising a expression construct comprising one or more nucleic
acids encoding an isolated protein complex according to the first-, second- or
third-mentioned aspect and a pharmaceutically-acceptable carrier or diluent.
In a sixth aspect, the invention provides a transformed cell capable
of expressing a recombinant protein complex, or recombinant proteins capable
of
forming said complex, according to the first-, second- or third-mentioned
aspects.
AMENDED SHEET
ippwat

CA 02424394 2003-03-21
PCT/AU01/01196
Received 21 August 2002
In a seventh aspect, the invention provides a method of modulating
cell proliferation and/or migration including the step of administering to an
animal
or isolated cells thereof, an isolated protein complex according to the first-
,
second- or third-mentioned aspects.
5 Preferably, the isolated protein complex comprises an IGFBP
and
vitronectin.
Preferably, the isolated protein complex further comprises IGF-I.
In an eighth aspect, the invention provides a method of modulating
cell proliferation and/or migration, including the step of administering to an
animal or isolated cells thereof an agent which prevents or disrupts formation
of a
protein complex according to the first-, second- or third-mentioned aspects.
Preferably, the agent prevents or disrupts an interaction between an
IGFBP and vitronectin.
More preferably, the agent prevents or disrupts an interaction
between IGFBP and vitronectin wherein the protein complex comprises IGF-I.
The agent may be an antagonist of an interaction between an
IGFBP and vitronectin or between an IGFBP-related protein and vitronectin, for
example.
An example of an agent that inhibits formation of IGFBP and
vitronectin complexes is IGF-11.
As will be described in more detail hereinafter, disruption of
interactions between vitronectin and IGFBPs may not only inhibit tumour cell
proliferation, but also inhibit tumour metastasis, both events being central
to
tumour pathology.
Further aspects of the invention provide uses of the isolated protein
complexes and methods according to the aforementioned aspects of the invention
in therapeutic or prophylactic treatments of diseases involving epithelial
cells such
as psoriasis, atherosclerosis, deterioration of the gastrointestinal
epithelium and
=
epithelial breast cancer, and/or in treatments which promote wound healing,
skin
repair, ulcer and bum healing, in vitro skin regeneration such as for grafting
of
autologous skin, bone regeneration and repair of damaged neuronal tissue.
Accordingly, the invention also provides a surgical implant or
AMEMED SHEET
IPF.A/AVI

CA 02424394 2003-03-21
1
PCT/AU01/01196
Received 21 August 2002
6
prosthesis comprising an isolated protein complex of the invention. The
surgical
implant or prosthesis may be coated, impregnated or otherwise pretreated with
said isolated protein complex.
The animal treated according to the invention may be a mammal,
preferably a human, or may be a non-mammalian vertebrate such as a fish,
reptile
or bird, or cells isolated from any of these.
Throughout this specification, unless otherwise indicated,
"comprise", "comprises" and "comprising" are used inclusively rather than
exclusively, so that a stated integer or group of integers may include one or
more
other non-stated integers or groups of integers.
BRIEF DESCRlPTION OF THE FIGURES AND TABLES
TABLE 1: List of references disclosing nucleic acids encoding growth
factors
and growth factor binding proteins.
TABLE 2: IGF-I and IGFBP-5 bound to vitronectin stimulates protein
synthesis in HaCAT human keratinocytes. Data are derived from measurements of
41-leucine incorporation and are expressed as % stimulation above control
(without IGF-I , IGFBP-5 or vitronectin) over 24 hr from a single experiment
in
which each treatment was tested in triplicate. IGF-I was added to IGFBP-5 (5
ng/well) in the presence (+) of vitronectin (300 ng/well) or in the absence (-
) of
vitronectin.
FIG. 1: Competition binding assay using increasing concentrations
of
either insulin (A) or IGF-11 (*)to compete with r251]-IGF-IE for binding to
300 ng
vitronectin (VN) per well. Radiolabeled IGF-IE (10,000 cpm) was added to VN-
coated wells and the number of counts bound was determined after overnight
incubation and several washes. The binding is expressed as a percentage of the
binding observed in control wells with no IGF-11 or insulin added. The results
are
shown as the average of three replicates standard deviation from a
representative
of three experiments.
FIG. 2: Competition binding assay using increasing concentrations
of
either IGF-I or IGF-IE to compete with [`251}-IGF-II for binding to 300 ng
vitronectin (VN) per well. Radiolabeled IGF-II (10,000 cpm) was added to
either
VN (M) - or IGFBP2 (*)-coated wells and the number of counts bound was
MENDED SHEET
'PENAL!

CA 02424394 2003-03-21
PCT/AU01/01196
Received 21 August 2002
7
determined after overnight incubation and several washes. The binding is
expressed as a percentage of the binding observed in control wells with no IGF
added.
FIG. 3:
Competition binding assay using increasing concentrations of
5. either
proIGF-II ( IP or IGF-II ( *) to compete with ['251]GF-II for binding to
300 ng vitronectin (VN) per well. Radiolabeled
(10,000 cpm) was added to
vitronectin-coated wells and the number of counts bound determined after an
overnight incubation and several washes. The binding is expressed as a
percentage
of the binding observed in the control wells with no added IGF-II or proIGF-E.
The results are, shown as the average of three replicates SEM from two
separate
experiments.
FIG. 4:
Competition binding assay using increasing concentrations of
either PM-1 OP or IGFAI (0) to compete with rINGF-II for binding to 300 ng
of vitronectin (VN) per well. Radiolabeled IGF-II (10,000 cpm) was added to
vitronectin-coated well and the number of counts bound was determined after an
overnight incubation and several washes. The binding is expressed as a
percentage
of the binding observed in the control wells with no added or
PM-i. The
results are shown as the average of three replicates standard deviation from
a
representative of three separate experiments.
FIG. 5: Competition
binding assay comparing effect of preincubation of
(A) and
IGFBP3 and (B) IGF-I and IGFBP3 upon binding to vitronectin.
IGFBP3 was non-glycosylated and produced in E. coli. Increasing concentrations
of IGFBP3 plus either 10,000 cpm ['251]GF-II (A) or ['25I]GF-I (B) were
preincubated for 4 hr and added to vitronectin-coated wells. Alternatively,
IGFBP3 plus either 10,000 cpm VINGF-II (A) or ['251]GF-I (B) were added to
vitronectin-coated wells without preincubation. The binding is expressed as
the
cpm obtained in the absence of non-specific binding. The results are shown as
the
average of three replicates from a representative of three separate
experiments.
FIG. 6:
Binding of labelled IGF-I to VN-coated wells in the presence of:
(A) IGFBP1, (B) IGFBP2, (C) IGFBP3, (D) IGFBP4, (E) IGFBP5 and (F)
IGFBP6. The recombinant IGFBPs were produced in mammalian cells. The data,
expressed as average cpm of labelled IGF-I bound/VN-coated well (300 ng/well)
AMENDED SHEET
IPENAU
=

CA 02424394 2003-03-21
PCT/AU01/01196
= Received 21 August 2002
8
in the presence of the indicated IGFBP are from six individual determinations.
Ten thousand cpm of radiolabelled IGF-I was added to each well.
FIG. 7:
Binding of labelled IGF-I to VN in the presence of "Gly IGFBP-3"
(glycoslyated IGFBP-3), "IGFBP-3 HBD mutant" (IGFBP-3 with the putative
heparin binding domain mutated) and to "non-gly IGFBP-3" (non-glycosylated
IGFBP-3).
FIG 8:
Competition binding assay using .increasing concentrations of IGFs
and desIGFs to compete with non-glycosylated IGFBP3 incubated with [12511-IGF-
I or
30 ng (A) or 10 ng (B) of IGFBP3 plus 10,000 cpm rINGF-IE (A) or
IGF-I (B) were added to VN-coated wells. Increasing concentrations of IGF-I,
IGF-11, des (1-3) IGF-I (not shown) and des(1-6) IGF-II were added to compete
with the radiolabel for binding to 'VN. Control wells coated with VN were
treated
with either 10 ng (A) or 30ng (B) IGFBP3 are shown together with control wells
coated with VN alone. The binding is expressed as cpm obtained in the absence
of
non-specific binding. The results are expressed as the average of three
replicates
standard deviation from a representative of three separate experiments.
FIG. 9:
Stimulation of protein synthesis in human keratinocytes. The data,
expressed as % stimulation above control (-VN, -IGF-I1) over 24 h is pooled
from
three replicate experiments in which each treatment was tested in triplicate.
The
theoretical additive effect represented by open bars (effect of VN alone)
combined
with grey bars (effect of IGF-II alone) is compared with the actual observed
effect
of IGF-IE prebound to 'VN (black bars). In all instances except the lowest
concentration of
tested, the actual observed effect is significantly greater (p
<0.05) than the calculated additive effect.
FIG. 10: Binding of
labelled IGF-II to 'VN-coated wells in the presence of:
(A) IGFBP1, (B) IGFBP2, (C) IGFBP3, (D) IGFBP4, (B) IGFBP5 and (F)
IGFBP6. The recombinant IGFBPs were produced in mammalian cells. The data,
expressed as average cpm of labelled IGF-11 bound/VN-coated well (300 ng/well)
in the presence of the indicated IGFBP are from six individual determinations.
Ten thousand cpm of radiolabelled was added to each well.
DETAILED DESCRIPTION OF THE INVENTION
The present invention has arisen, at least in part, from the discovery
Aikli ENDED allEET

CA 02424394 2003-03-21
PCT/AU01/01196
Received 21 August 2002
9
by the present inventors that IGF-I binds vitronectin via a binding
interaction
between an IGFBP and vitronectin. Furthermore, the present invention describes
variant IGFs and IGFBPs that may be used to augment or diminish binding
between IGFS, IGFBPs and vitronectin. These discoveries have led the present
inventors to manipulate these binding interactions in vitro with a view to
manipulating contingent in vivo biological events associated with cell growth,
proliferation and migration. This invention therefore has utility in medical
treatments such as wound healing, skin repair and maintenance, bone
regeneration, atherosclerosis and cancer therapy in both medical and
veterinary
areas.
For the purposes of this invention, by "isolated" is meant removed
from a natural state or otherwise subjected to human manipulation. Isolated
material may be substantially or essentially free from components that
normally
accompany it in its natural state, or may be manipulated so as to be in an
artificial
state together with components that normally accompany it in its natural
state.
By "polypeptide" is also meant "protein", either term referring to
an amino acid polymer.
Proteins and peptides inclusive of growth factor, growth factor
binding proteins and vitronectin proteins may be isolated in native, chemical
synthetic or recombinant synthetic form.
A "peptide" is a protein having no more than fifty (50) amino
acids.
A "biologically-active fragment" is a fragment, portion or segment
of a protein which displays at least 1%, preferably at least 10%, more
preferably at
least 25% and even more preferably at least 50% of the biological activity of
the
protein.
Peptides may be readily synthesized by recombinant or chemical
synthesis techniques. For example, reference may be made to solution synthesis
or
solid phase synthesis as described, for example, in Chapter 9 entitled
"Peptide
Synthesis" by Atherton and Shephard which is included in a publication
entitled
"Synthetic Vaccines" edited by Nicholson and published by Blackwell Scientific
Publications. Peptide synthesis methods are also described in Chapter 18 of
AliENDED SHEET
112=aiDi

CA 02424394 2010-05-31
CURRENT PROTOCOLS IN PROTEIN SCIENCE Eds. Coligan et al. (John Wiley & Sons
NY, 1997). Alternatively, peptides can be produced by digestion of a
polypeptide of the
invention with proteinases such as endoLys-C, endoArg-C, endoGlu-C and
staphylococcus V8-
protease. The digested fragments can be purified by, for example, high
performance liquid
5 chromatographic (HPLC) techniques.
In a preferred form, the invention provides an isolated protein complex
comprising
vitronectin, a growth factor and a growth factor binding protein.
By "growth factor" is meant a molecule that stimulates or promotes growth of
an
organism or cells of said organism. A preferred growth factor is a protein or
peptide that
10 stimulates cell division, and in particular, mammalian cell division.
Preferably, isolated protein complexes of the invention comprise the growth
factor IGF-I.
Isolated IGF and IGFBP polypeptides are commercially available from sources
such as
GroPep (Adelaide, Australia), while VN polypeptides are commercially available
from sources
such as Promega Corporation (Madison WI USA). Recombinant IGFs, IGFBPs and VN
are
readily made by persons skilled in the art, as will be discussed in more
detail hereinafter.
As will be appreciated by the skilled persons, the invention also includes
precursor forms
of IGFs. Examples of pro-IGF-I proteins are IGF-I proteins that have had the
signal peptide
removed but are not fully processed by cleavage of the E-domain. IGF-I has
three different E
domains that result from differential mRNA splicing and these precursor
proteins may be
present in isolated protein complexes of the invention.
However, the invention also contemplates isolated protein complexes comprising
a
growth factor or other biologically active protein such as epidermal growth
factor (EGF; (Heldin
et al., 1981, Science 4 1122-1123), fibroblast growth factor (FGF; Nurcombe et
al., 2000, J.
Biol. Chem. 275 30009-30018), basic fibroblast growth factor (bFGF;
Taraboletti et al., 1997,
Cell

CA 02424394 2003-03-21
PUT/AU01/01196
Received 21 August 2002
Growth. Differ. 8 471-479), osteopontin (Nam et aL, 2000, Endocrinol. 141
1100), thrombospondin-1 (Nam et al., 2000, supra), tenascin-C (Arai et al.,
1996,
J. Biol. Chem. 271 6099), PM-1 (Nam et al., 1997, Endocrinol. 138 2972),
plasminogen (Campbell et al., 1998, Am. J. Physiol. 275 E321), fibrinogen
(Campbell et al., 1999, J. Biol. Chem 274 30215), fibrin (Campbell et al.,
1999,
supra) or transferrin (Weinzimer et al., 2001, J. Clin. Endocrinol. Metab. 86
1806).
Preferably, isolated protein complexes comprising osteopontin
thrombospondin-1, tenascin-C or PM-1 further comprise IGFBP-5.
Preferably, isolated protein complexes comprising plasminogen
fibrinogen, fibrin or transferrin further comprise IGFBP-3.
The invention contemplates isolated protein complexes comprising
monomeric and multimeric vitronectin, as vitronectin can exist in monomeric
and
mutimeric states. In particular, multimeric VN accumulates in areas of
vascular
injury and also is the predominant form of VN in tissue. Thus the multimeric
form
of VN provides the opportunity to form a VN complex in which more than one
type of growth factor or growth factor binding protein can be delivered at the
same
time.
= It will also be appreciated by the skilled person that isolated protein
complexs of the invention may include vitronectin in "native", "denatured" or
"extended" states as are well undestood in the art.
The invention also contemplates isolated growth factor complexes
comprising nectinepsin, which is an extracellular matrix protein that shows
60%
homology to VN at the amino acid level (Blanchert et al., 1996, J. Biol. Chem.
271 26220-26226).
It will also be understood that variants of growth factors, growth
factor binding proteins and/or vitronectin may be used to form isolated
protein
complexes of the invention and may be useful in the methods of use set forth
herein.
As used herein, "variant" proteins, polypeptides and peptides of the
invention include those in which one or more amino acids have been replaced by
AMENDED SHEET
IPE.M/AU

CA 02424394 2003-03-21
PCT/AU01/01196
Received 21 August 2002
ha
different amino acids.
It is well understood in the art that some amino acids may be
changed to others with broadly similar properties without changing the nature
of
MENDED EHET
IPE*4)

CA 02424394 2003-03-21
WO 02/24219 PCT/AU01/01196
12
the activity of the polypeptide (conservative substitutions).
Substantial changes in function may be made by selecting
substitutions that are less conservative. Other replacements would be non-
conservative substitutions and relatively fewer of these may be tolerated.
Generally, the substitutions which are likely to produce the greatest changes
in a
polypeptide's properties are those in which (a) a hydrophilic residue (e.g.,
Ser or
Thr) is substituted for, or by, a hydrophobic residue (e.g., Ala, Leu, Ile,
Phe or
Val); (b) a cysteine or proline is substituted for, or by, any other residue;
(c) a
residue having an electropositive side chain (e.g., Arg, His or Lys) is
substituted
for, or by, an electronegative residue (e.g., Glu or Asp) or (d) a residue
having a
bulky side chain (e.g., Phe or Trp) is substituted for, or by, one having a
smaller
side chain (e.g., Ala, Ser)or no side chain (e.g., Gly).
Variants also include proteins, polypepfides and peptides which
have been altered, for example by conjugation or complexing with other
chemical
moieties or by post-translational modification techniques as would be
understood
in the art. Such derivatives include amino acid deletions and/or additions.
Other polypeptide and peptide variants contemplated by the
invention include, but are not limited to, modification to side chains,
incorporation
of unnatural amino acids and/or their derivatives during peptide, polypeptide
or
2 0 protein synthesis and the use of crosslinkers and other chemicals which
impose
conformational constraints on the polypeptides and peptide variants of the
invention. Examples of side chain modifications contemplated by the present
invention include modifications of amino groups such as by acylation with
acetic
anhydride; acylation of amino groups with succinic anhydride and
tetrahydrophthalic anhydride; amidination with methylacetimidate;
carbamoylation of amino groups with cyanate; pyridoxylation of lysine with
pyridoxa1-5-phosphate followed by reduction with NaBH4; reductive alkylation
by
reaction with an aldehyde followed by reduction with NaBH4; and
trinitrobenzylation of amino groups with 2, 4, 6-trinitrobenzene sulphonic
acid
(TNBS).
The carboxyl group may be modified by carbodiimide activation
via 0-acylisourea formation followed by subsequent derivitization, by way of

CA 02424394 2003-03-21
WO 02/24219 PCT/AU01/01196
13
example, to a corresponding amide.
The guanidine group of arginine residues may be modified by
formation of heterocyclic condensation products with reagents such as 2,3-
butanedione, phenylglyoxal and glyoxal.
Sulphydryl groups may be modified by methods such as performic
acid oxidation to cysteic acid; formation of mercurial derivatives using 4-
chloromercuriphenylsulphonic acid, 4-chloromercuribenzoate; 2-chloromercuri-4-
nitrophenol, phenylmercury chloride, and other mercurials; folination of a
mixed
disulphides with other thiol compounds; reaction with maleimide, maleic
anhydride or other substituted maleimide; carboxymethylation with iodoacetic
acid or iodoacetamide; and carbamoylation with cyanate at alkaline pH.
Tryptophan residues may be modified, for example, by alkylation
of the indole ring with 2-hydroxy-5-nitrobenzyl bromide or sulphonyl halides
or
by oxidation with N-bromosuccinimide.
Tyrosine residues may be modified by nitration with
tetranitromethane to form a 3-nitrotyrosine derivative.
The imidazole ring of a histidine residue may be modified by N-
carbethoxylation with diethylpyrocarbonate or by alkylation with iodoacetic
acid
derivatives.
Examples of incorporating unnatural amino acids and derivatives
during peptide synthesis include but are not limited to, use of 4-amino
butyric
acid, 6-aminohexanoic acid, 4-amino-3-hydroxy-5-phenylpentanoic acid, 4-
amino-3-hydroxy-6-methylheptanoic acid, t-butylglycine, norleucine, norvaline,
phenylglycine, ornithine, sarcosine, 2-thienyl alanine and/or D-isomers of
amino
acids.
Modifications also include within their scope 0- and N- linked
glycosylation variants and non-glycosylated forms of proteins that, in their
naturally-occurring state, are glycosylated.
With regard to variants, these may be created by mutagenizing a
polypeptide or by mutagenizing an encoding nucleic acid, such as by random
mutagenesis or site-directed mutagenesis. Examples of nucleic acid mutagenesis
methods are provided in in Chapter 9 of CURRENT PROTOCOLS IN

CA 02424394 2010-05-31
14
MOLECULAR BIOLOGY, Ausubel et al., supra.
It will be appreciated by the skilled person that site-directed mutagenesis is
best
performed where knowledge of the amino acid residues that contribute to
biological activity is
available. In many cases, this information is not available, or can only be
inferred by molecular
modelling approximations, for example.
In such cases, random mutagenesis is contemplated. Random mutagenesis methods
include chemical modification of proteins by hydroxylamine (Ruan et al., 1997,
Gene 188 35),
incorporation of dNTP analogs into nucleic acids (Zaccoto et al., 1996, J.
Mol. Biol. 255 589)
and PCR-based random mutagenesis such as described in Stemmer, 1994, Proc.
Natl. Acad. Sci.
USA 91 10747 or Shafikhani et al., 1997, Biotechniques 23 304. It is also
noted that PCR-based
random mutagenesis kits are commercially available, such as the DiversifyTM
kit (Clontech).
The invention also contemplates use of growth factor variants such as des(1-
6)IGF-II and
des(1-3)IGF-I to form isolated protein complexes of the invention.
Other variant 1GFs and IGFBPs useful as agonists or antagonists will be
described in
more detail hereinafter.
Recombinant growth factor complexes
It will be appreciated that isolated protein complexes may be produced using
recombinant growth factors, growth factor binding proteins and/or vitronectin,
by expression of
an encoding nucleic acid in an appropriate host cell or in a cell-free
expression system as are
well known in the art.
The term "nucleic acid" as used herein designates single-or double-stranded
mRNA,
RNA, cRNA and DNA, said DNA inclusive of cDNA and genomic DNA.
A "polynucleotide" is a nucleic acid having eighty (80) or more contiguous
nucleotides,
while an "oligonucleotide" has less than eighty (80) contiguous nucleotides.

CA 02424394 2010-05-31
A "probe" may be a single or double-stranded oligonucleotide or
polynucleotide, suitably
labeled for the purpose of detecting complementary sequences in Northern or
Southern blotting,
for example.
A "primer" is usually a single-stranded oligonucleotide, preferably having 15-
50
5
contiguous nucleotides, which is capable of annealing to a complementary
nucleic acid
"template" and being extended in a template-dependent fashion by the action of
a DNA
polymerase such as Taq polymerase, RNA-dependent DNA polymerase or
SequenaseTM.
Nucleic acids encoding IGFs, EGF, IGFBPs, ALS and VN are well known in the art
and
have been available for many years. However, the skilled person is referred to
Table 1 which
10 lists references that provide examples of these nucleic acid sequences.
Nucleic acids useful according to the present invention may be prepared
according to the
following procedure:
(i)
creating primers which are, optionally, degenerate wherein each comprises a
respective portion of a target nucleic acid; and
15 (ii)
using said primers in combination with a nucleic acid amplification technique
to
amplify one or more amplification products from a nucleic acid extract.
Suitable nucleic acid amplification techniques are well known to the skilled
person, and
include polymerase chain reaction (PCR) as for example described in Chapter 15
of Ausubel et
al. supra; strand displacement amplification (SDA) as for example described in
U.S. Patent No
5,422,252; rolling circle replication (RCR) as for example described in Liu et
al., 1996, J. Am.
Chem. Soc. 118 1587, International application WO 92/01813 and International
Application
WO 97/19193; nucleic acid sequence-based amplification (NASBA) as for example
described
by Sooknanan et al., 1994, Biotechniques 17 1077; ligase chain

CA 02424394 2010-05-31
16
reaction (LCR) as for example described in International Application
W089/09385; and Q-13
replicase amplification as for example described by Tyagi et al., 1996, Proc.
Natl. Acad. Sci.
USA 93 5395.
As used herein, an "amplification product" refers to a nucleic acid product
generated by
nucleic acid amplification techniques.
Recombinant proteins may be prepared by any suitable procedure known to those
of skill
in the art.
For example, the recombinant protein may be prepared by a procedure including
the
steps of:
(i) preparing an expression construct which comprises a nucleic acid,
operably
linked to one or more regulatory nucleotide sequences;
(ii) transfecting or transforming a suitable host cell with the expression
construct; and
(iii) expressing the polypeptide in said host cell.
For the purposes of host cell expression, the recombinant nucleic acid is
operably linked
to one or more regulatory sequences in an expression vector.
An "expression vector" may be either a self-replicating extra-chromosomal
vector such
as a plasmid, or a vector that integrates into a host genome.
By "operably linked" is meant that said regulatory nucleotide sequence(s)
is/are
positioned relative to the recombinant nucleic acid of the invention to
initiate, regulate or
otherwise control transcription.
Regulatory nucleotide sequences will generally be appropriate for the host
cell used for
expression. Numerous types of appropriate expression vectors and suitable
regulatory
sequences are known in the art for a variety of host cells.
Typically, said one or more regulatory nucleotide sequences may include, but
are not
limited to, promoter sequences, leader or signal sequences,

CA 02424394 2003-03-21
WO 02/24219 PCT/AU01/01196
17
ribosomal binding sites, transcriptional start and termination sequences,
translational start and termination sequences, and enhancer or activator
sequences.
Constitutive or inducible promoters as known in the art are
contemplated by the invention. The promoters may be either naturally occurring
promoters, or hybrid promoters that combine elements of more than one
promoter.
In a preferred embodiment, the expression vector contains a
selectable marker gene to allow the selection of transformed host cells.
Selectable
marker genes are well known in the art and will vary with the host cell used.
The expression vector may also include a fusion partner (typically
provided by the expression vector) so that the recombinant polypeptide of the
invention is expressed as a fusion polypeptide with said fusion partner. The
main
advantagse of fusion partners are that they assist identification and/or
purification
of said fusion polypeptide and also enhance protein expression levels and
overall
yeild.
In order to express said fusion polypeptide, it is necessary to ligate
a nucleotide sequence according to the invention into the expression vector so
that
the translational reading frames of the fusion partner and the nucleotide
sequence
of the invention coincide.
Well known examples of fusion partners include, but are not
limited to, glutathione-S-transferase (GST), Fc potion of human IgG, maltose
binding protein (MBP) and hexahistidine (HIS6), which are particularly useful
for
isolation of the fusion polypeptide by affinity chromatography. For the
purposes
of fusion polypeptide purification by affinity chromatography, relevant
matrices
for affinity chromatography are glutathione-, amylose-, and nickel- or cobalt-
2 5 conjugated resins respectively. Many such matrices are available in
"kit" form,
such as the QlAexpressTM system (Qiagen) useful with (HIS6) fusion partners
and
the Pharmacia GST purification system.
Another fusion partner well known in the art is green fluorescent
protein (GFP). This fusion partner serves as a fluorescent "tag" which allows
the
fusion polypeptide of the invention to be identified by fluorescence
microscopy or
by flow cytometry. The GFP tag is useful when assessing subcellular
localization
of the fusion polypeptide of the invention, or for isolating cells which
express the

CA 02424394 2010-05-31
18
fusion polypeptide of the invention. Flow cytometric methods such as
fluorescence activated
cell sorting (FACS) are particularly useful in this latter application.
In some cases, the fusion partners also have a protease cleavage site, such as
for Factor
Xa or Thrombin, which allow the relevant protease to partially digest the
fusion polypeptide of
the invention and thereby liberate the recombinant polypeptide of the
invention therefrom. The
liberated polypeptide can then be isolated from the fusion partner by
subsequent
chromatographic separation.
Fusion partners according to the invention also include within their scope
"epitope tags",
which are usually short peptide sequences for which a specific antibody is
available. Well
known examples of epitope tags for which specific monoclonal antibodies are
readily available
include c-myc, influenza virus haemagglutinin and FLAG tags.
The recombinant protein may be conveniently prepared by a person skilled in
the art
using standard protocols as for example described in Sambrook, et al.,
MOLECULAR
CLONING. A Laboratory Manual (Cold Spring Harbor Press, 1989), in particular
Chapter 16
and 17; CURRENT PROTOCOLS IN MOLECULAR BIOLOGY Eds. Ausubel et al., (John
Wiley & Sons, Inc. 1995-1999), in particular Chapters 10 and 16; and CURRENT
PROTOCOLS IN PROTEIN SCIENCE Eds. Coligan et al., (John Wiley & Sons, Inc.
1995-
1999), in particular Chapters 1, 5 and 6.
In one embodiment, recombinant expression of growth factor, growth factor
binding
protein and vitronectin may he performed separately, and complexes formed
therefrom.
In another embodiment, recombinant expression of growth factor, growth factor
binding
protein and vitronectin may be performed in the same cell, and complexes
formed therefrom.
As hereinbefore, polypeptides of the invention may be produced by culturing a
host cell
transformed with said expression construct comprising a nucleic acid encoding
a polypeptide, or
polypeptide homolog, of the invention.

CA 02424394 2010-05-31
19
The conditions appropriate for protein expression will vary with the choice of
expression vector
and the host cell. This is easily ascertained by one skilled in the art
through routine
experimentation.
Suitable host cells for recombinant expression include bacteria such as E.
coli.,
Clostridium sp., Pseudomonas sp., yeast, plant cells, insect cells (such as
SP) and mammalian
cells such as fibroblasts and keratinocytes.
Preferred host cells are human keratinocytes.
Inducible and non-inducible expression vectors are contemplated. In stably
transfected
mammalian cells, a number of inducible and repressible systems have been
devised including
metallothionine (MT) inducible and tetracycline and repressible (tetR), each
of which is
contemplated by the present invention.
Particular examples of suitable expression vectors and methods of recombinant
IGFBP
expression may be found in United States Patent 5,973,115.
Mimetics, Agonists and Antagonists
The invention contemplates agents which may promote, prevent or disrupt
formation of
protein complexes comprising growth factors, growth factor binding proteins
and vitronectin.
Such an agent may be a mimetic. The term "mimetic" is used herein to refer to
molecules that
are designed to resemble particular functional regions of proteins or
peptides, and includes
within its scope the terms "agonist", "analogue" and "antagonist" as are well
understood in the
art.
Of relevance is the elucidation of the portions of IGFs and IGFBPs which are
responsible
for IGF-IGFBP binding, as described in International Publication W000/23469.
Furthermore,
agonist variants of IGF-I have been made which selectively bind IGFBP-1 or
IGFBP-3, as
described in International Publication W000/40612.
It is therefore contemplated that agents could be engineered which disrupt or
prevent
formation of polypeptide complexes between IGFBPs and VN. An example would be
a peptide
which competes for binding of the IGFBP to VN by resembling the binding site
on VN or the
IGFBP.

CA 02424394 2003-03-21
PUT/AU01/01196
= Received 18 April 2002
As will be described in more detail hereinafter, is
an agent
that can inhibit binding between an IGFBP and vitronectin. It is also proposed
that
residues V35 or S36 along with S" of the RVSRRSR sequence at positions 34-40
in
the C-domain of IGF-II could be deleted to thereby resemble the HBD of IGFBP3
5 (BXBBB
wherein B is a basic amino acid residue). This would create an agent
even more capable of inhibiting formation of a complex between IGFBPs and
vitronectin.
An example of an agonist contemplated by the present invention is
IGF-1 engineered to include a heparin binding domain (HBD) of an IGFBP to
10
thereby bind vitronectin directly. For example, the sequence SSSRRAPQT in the
C-domain of IGF-I may be engineered to have a BXBBB motif. A preferred
BXBBB motif is the KGRKR sequence of IGFBP-3 (residues 228-232).
Alternatively, the putative vitronectin-binding domain of IGF-1:1:
RVSRRSR (residues 34-40) may be introduced into IGF-I.
15 An
example of an antagonist of the invention is an IGFBP
engineered to mutate basic residues in the HBD (as hereinbefore described) to
reduce or prevent binding of the IGFBP to vitronectin.
Suitably, the engineered IGFBP is capable of binding IGF-I.
Preferably, the engineered IGFBP is IGFBP-3 or IGFBP-5.
20 It is
also contemplated that an analogue of an IGFBP could be
engineered which enables formation of a complex between the analogue and VN.
Suitably, the analogue would also bind an IGF. Potential advantages of such an
analogue is that it might be more readily synthesized or isolated than an
IGFBP,
have a particular desired biological half-life and perhaps be engineered to
specifically bind IGF-I.
The aforementioned mimetics may be peptides, polypeptides or
other organic molecules, preferably small organic molecules, with a desired
biological activity and half-life.
Computer-assisted structural database searching is becoming
increasingly utilized as a procedure for identifying mimetics.. Database
searching
methods which, in principle, may be suitable for identifying mimetics, may be
found in International Publication WO 94/18232 (directed to producing HIV
AMENDED SHEET
!MAW

CA 02424394 2010-05-31
21
antigen mimetics), United States Patent No, 5,752,019 and International
Publication WO
97/41526 (directed to identifying EPO mimetics).
Other methods include a variety of biophysical techniques which indentify
molecular
interactions. These allow for the screening of candidate molecules according
to whether said
candidate molecule affects formation of IGF-IGFBP-VN complexes, for example.
Methods
applicable to potentially useful techniques such as competetive radioligand
binding assays (see
Upton et al., 1999, supra for a relevant method), analytical
ultracentrifugation,
microcalorimetry, surface plasmon resonance and optical bio sensor-based
methods are provided
in Chapter 20 of CURRENT PROTOCOLS IN PROTEIN SCIENCE Eds. Coligan et al.,
(John
Wiley & Sons, 1997).
Pharmaceutical compositions
The invention includes administration of a protein complex of the invention in
the form
of a pharmaceutical composition. Pharmaceutical compositions of the invention
may include
isolated protein complexes that comprise variant IGFs and/or IGFBPs or agents
that disrupt or
prevent formation of said complexes as hereinbefore described.
Suitably, the pharmaceutical composition comprises a pharmaceutically-
acceptable
carrier. Pharmaceutical compositions may also include polypeptide variants,
fragments or
mimetics as hereinbefore defined.
By "pharmaceutically-acceptable carrier" is meant a solid or liquid filler,
diluent or
encapsulating substance that may be safely used in systemic administration.
Depending upon
the particular route of administration, a variety of carriers, well known in
the art may be used.
These carriers may be selected from a group including sugars, starches,
cellulose and its
derivatives, malt, gelatine, talc, calcium sulfate, vegetable oils, synthetic
oils, polyols, alginic
acid, phosphate buffered solutions, emulsifiers, isotonic saline, and pyrogen-
free water.
Any suitable route of administration may be employed for providing a patient
with the
composition of the invention. For example, oral, rectal, parenteral,
sublingual, buccal,
intravenous, intra-articular, intra-muscular,

CA 02424394 2010-05-31
22
intra-dermal, subcutaneous, inhalational, intraocular, intraperitoneal,
intracerebroventricular,
transdermal and the like may be employed. Intra-muscular and subcutaneous
injection is
appropriate, for example, for administration of immunogenic compositions,
vaccines and DNA
vaccines.
Dosage forms include tablets, dispersions, suspensions, injections, solutions,
syrups,
troches, capsules, suppositories, aerosols, transdermal patches and the like.
These dosage forms
may also include injecting or implanting controlled releasing devices designed
specifically for
this purpose or other forms of implants modified to act additionally in this
fashion. Controlled
release of the therapeutic agent may be effected by coating the same, for
example, with
hydrophobic polymers including acrylic resins, waxes, higher aliphatic
alcohols, polylactic and
polyglycolic acids and certain cellulose derivatives such as
hydroxypropylmethyl cellulose. In
addition, the controlled release may be effected by using other polymer
matrices, liposomes
and/or microspheres.
Pharmaceutical compositions of the present invention suitable for oral or
parenteral
administration may be presented as discrete units such as capsules, sachets or
tablets each
containing a pre-determined amount of one or more therapeutic agents of the
invention, as a
powder or granules or as a solution or a suspension in an aqueous liquid, a
non-aqueous liquid,
an oil-in-water emulsion or a water-in-oil liquid emulsion.
With regard to pharmaceutical compositions comprising IGF and IGFBPs,
particular
reference is made to U.S. patent 5,936,064 and International Publications
W099/62536 and
W099/54359.
Pharmaceutical compositions of the invention may also include expression
vectors such
as viral vectors such as vaccinia, and viral vectors useful in gene therapy.
The latter include
adenovirus and adenovirus-associated viruses (AAV) such as described in Braun-
Falco et al.,
1999, Gene Ther. 6 432, retroviral and lentiviral vectors such as described in
Buchshacher et al.,
2000, Blood 95 2499 and vectors derived from herpes simplex virus and
cytomegalovirus. A

CA 02424394 2010-05-31
23
general overview of viral vectors useful in endocrine gene therapy is provided
in Stone et al.,
2000, J. Endocrinol. 164 103.
The present invention may also utilize specific expression vectors which
target gene
expression to epidermal cells, such as described in United States Patent
5,958,764 and for in
vivo wound healing applications, such as described in United States Patent
5,962,427.
Therapeutic uses
The invention provides methods of treatment using polypeptide complexes of the
invention. These methods are particularly aimed at therapeutic treatment of
mammals, and more
particularly, humans.
Such methods include administration of pharmaceutical compositions as
hereinbefore
defined, and may be by way of microneedle injection into specific tissue
sites, such as described
in U.S. patent 6,090,790, topical creams, lotions or sealant dressings applied
to wounds, burns or
ulcers, such as described in U.S. patent 6,054,122 or implants which release
the composition
such as described in International Publication W099/47070.
Gene therapy is also applicable in this regard, such as according to methods
set forth in
United States Patent 5,929,040 and United States Patent 5,962,427.
There also exist methods by which skin cells can be genetically modified for
the purpose
of creating skin substitutes, such as by genetically engineering desired
growth factor expression
(Supp et al., 2000, J. Invest. Dermatol. 114 5). An example of a review of
this field is provided
in Bevan et al., Biotechnol. Gent. Eng. Rev. 16 231.
Also contemplated is "seeding" a recipient with transfected or transformed
cells, such as
described in International Publication W099/11789.
These methods can be used to stimulate cell proliferation and thereby
facilitate or
progress wound and burn healing, repair of skin lesions such as ulcers, tissue
replacement and
grafting such as by in vitro culturing of

CA 02424394 2003-03-21
WO 02/24219 PCT/AU01/01196
24
autologous skin, re-epithelialization of internal organs such as kidney and
lung
and repair of damaged nerve tissue.
Skin replacement therapy has become well known in the art, and
may employ use of co-cultured epithelial/keratinocyte cell lines, for example
as
described in Kehe et al., 1999, Arch. Dermatol. Res. 291 600 or in vitro
culture of
primary (usually autologous) epidermal, dermal and/or keratinocyte cells.
These
techniques may also utilize engineered biomaterials and synthetic polymer
"scaffolds".
Examples of reviews of the field in general are provided in
Terskikh & Vasiliev, 1999, Int. Rev. Cytol. 188 41 and Eaglestein & Falanga,
1998, Cutis 62 1.
More particularly, the production of replacement oral mucosa
useful in craniofacial surgery is described in Izumi et al., 2000, J. Dent.
Res. 79
798. Fetal keratinocytes and dermal fibroblasts can be expanded in vitro to
produce skin for grafting to treat skin lesions, such as described in Fauza et
al., J.
Pediatr. Surg. 33 357, while skin substitutes from dermal and epidermal skin
elements cultured in vitro on hyaluronic acid-derived biomaterials have been
shown to be potentially useful in the treatment of burns (Zacchi et al., 1998,
J.
Biomed. Mater. Res, 40 187).
Another aspect of epithelial cell therapy relates to healing of the
epithelial lining of the gastrointestinal tract to treat or prevent impaired
gut
function.
Polymer scaffolds are also contemplated for the purpose of
facilitating replacement skin engineering, as for example described in
Sheridan et
al., 2000, J. Control Release 14 91 and Fauza et al., 1998, .supra, as are
microspheres as agents for the delivery of skin cells to wounds and burns
(LaFrance & Armstrong, 1999, Tissue Eng. 5 153).
The aforementioned techniques may be readily utilized according
to the present invention by use of isolated protein complexes of the invention
to
promote skin cell proliferation for the purposes of tissue replacement and for
cosmetic skin treatments.

CA 02424394 2003-03-21
WO 02/24219 PCT/AU01/01196
With regard to bone regeneration, the invention provides surgical
or prosthetic implants coated, impregnated or otherwise pretreated with an
isolated protein complex of the invention.
Conversely, inhibition or suppression of cell proliferation and
5 migration by preventing or disrupting formation of IGF-IGFBP-VN
complexes
may constitute a prophylactic or therapeutic treatment of psoriasis or
malignancies
such as epithelial cancers such as breast cancer.
The invention also contemplates a method of differentiating a stem
or progenitor cell by administering an isolated protein complex of the
invention to
10 said stem or progenitor cell. Isolated protein complexes comprising
variant growth
factors and IGFBPs may also be applicable to this method.
Differentiated cells produced according to this method may be
useful in therapeutic methods such as as hereinbefore described.
For example, smooth muscle cells are considered to be
15 "mesenchymal stem cells" and may be driven to differentiate into
fibroblasts,
stromal cells, endothelial cells, bone or adipocytes.
In order that the invention may be readily understood and put into
practical effect, particular preferred embodiments will now be described by
way of
the following non-limiting examples.
2 0 All competitive radioligand binding assays as described herein
were performed essentially as described in Upton et al., 1999, supra.
EXAMPLE 1
Competition binding assay to assess ability of insulin, pro-IGF-H and IGF-I to
compete with IGF-H for binding to vitronectin
25 Due to the similarity in structure shared between IGFs and insulin,
binding of
insulin to vitronectin was examined. Crosslinking experiments performed by
Upton et al., 1999, supra indicated that insulin was unlikely to compete with
IGF-11 for binding to vitronectin, as was the case with IGF-I. As a
consequence,
high concentrations of insulin were examined in order to establish whether
insulin
could compete with radiolabelled IGF-11 for binding to vitronectin. The
results
shown in FIG.1 indicated that, as for the situation originally observed with
IGF-I

CA 02424394 2003-03-21
WO 02/24219 PCT/AU01/01196
26
(Upton et al., 1999, supra), insulin competed poorly with [1251]-IGF-11 for
binding
to vitronectin.
Investigation of IGF-I as a competitor for IGF-II binding to
vitronectin revealed that IGF-I, could compete with the binding of [125I]-IGF-
11 to
vitronectin (FIG. 2). However, IGF-I was markedly less effective than IGF-II
in
competing with radiolabelled IGF-11 for binding to vitronectin, requiring an
approximate 3000 fold increase in IGF-I concentration above IGF-II to achieve
the
same effect.
Similar studies demonstrated that proIGF-II could compete with
[125I]-IGF-II for binding to vitronectin (FIG. 3). ProIGF-II was not as
effective as
IGF-II in competing with ['251]-IGF-11 for binding to vitronectin with IC50
values
of 65.6 nM and 9.6 nM respectively. Thus, the presence of the E domain within
proIGF-II represents a structural modification that alters the binding to
vitronectin.
This could be due to steric hindrance by the additional domain in proIGF-II
compared to IGF-ll, or may be a result of an alterted structure of the proIGF-
11
molecule that alters the affinity of proIGF-II for vitronectin.
EXAMPLE 2
Investigation of PAI-1 and uPAR as competitors for binding of IGF-II to
vitronectin
PAI-1 and suPAR were investigated as molecules which might compete with
IGF-II for binding to vitronectin, as these proteins have been reported to
bind
vitronectin (Declerck et al., 1988, J. Biol. Chem. 263 15454; Wei et a/.
,1994, J.
Biol. Chem. 269 32380; Kanse et a/.,1996, Exp. Cell Res. 224 344). At
concentrations up to 2000 nM competition of PAI-1 with 1251-IGF-II for binding
to
vitronectin was observed with an approximate IC50 value of 524 nM (FIG. 4).
While this concentration is relatively high in terms of IC50 values, such
concentrations can be found in vivo associated with tumours (Grondahl-Hansen
et
al.,1993, Cancer Res. 53 2513). Indeed, Kjoller et al.,1997, Exp. Cell Res.
232
420, found 370 nM of PAI-1 was required to achieve half maximal effect in an
3 0 assay assessing the ability of PAI-1 to inhibit cell migration of WISH
cells. This
inhibition was postulated to result via PM-1 competing with integrins and uPAR
for binding to vitronectin. Thus, the observed interaction between PAI-I, IGF-
II

CA 02424394 2003-03-21
WO 02/24219 PCT/AU01/01196
27
and vitronectin may indeed have some functional consequences in vivo,
particularly when IGF-II and PM-1 levels are highly expressed as is sometimes
found in tumours.
While it is known that PAI-1 binds to the N-terminal somatomedin
B domain of vitronectin, the high concentrations of PAT-1 required to
effectively
compete with for
binding to vitronectin suggested that these studies have
not provided any clear information regarding the location of of
the binding
site on vitronectin. However, the results may be interpreted two ways. The
first
possibility is that the observed competition between and
PAI-1 is due to
partial competition at the primary high affinity site on vitronectin within
the
somatomedin B domain via steric hindrance. Alternatively, the competition
could
be due to direct competition between and
PAI-1, for binding to a site on
vitronectin to which PM-1 binds with a reduced affinity. Further experimental
studies are required to clarify this issue.
While the present inventors were unable to show that soluble
suPAR competes for binding of IGF-II to VN, this may have been due to that
fact
that lyophilized suPAR was used after international transportation. There is
no
evidence that reconstituted suPAR is biologically active after reconstitution,
although the observation that P511-suPAR did not bind VN (data not shown)
perhaps supports an interpretation that the suPAR used in these studies was
inactive.
Use of similar techniques to those that identified the PM-1 binding
site on vitronectin to be within a fragment of vitronectin containing the 44
N-terminal amino acids (Deng et al., 1995, Thromb. Haemost. 74 66), may
establish whether the competition observed between PM-1 and IGF-II for binding
vitronectin is a result of either of these possibilities.
It has previously been shown that the soluble form of the urokinase
receptor (suPAR) can bind to immobilized vitronectin (Wei et al., 1994,
supra). In
the studies reported here, however, even at concentrations up to 300 nM, no
competition between suPAR and ['251]GF-II for binding to vitronectin was
observed.
EXAMPLE 3

CA 02424394 2003-03-21
WO 02/24219 PCT/AU01/01196
28
Binding of IGF-I and IGF-II to vitronectin in the presence of non-glycosylated
recombinant IGFBP-3
To investigate whether IGFBPs could mediate binding of the IGFs to
vitronectin,
the effect of increasing concentrations of IGFBP-3 in the presence of {'251]GF-
I
or ['251]GF-II was assessed. The results are shown in FIG. 5.
At the concentrations tested, 10 ng of IGFBP-3 per well caused the
greatest amount of [125I3-IGF-I to bind to vitronectin coated wells, whereas
30 ng
of IGFBP-3 per well caused the greatest amount of [1251]GF-I1 to bind to
vitronectin coated wells. The effect was most noticeable in the case of
['251]GF-I,
since low counts of [125INGF-I bound to vitronectin, and even at the lowest
concentration of IGFBP-3 (3.7 ng), this binding was increased (FIG. 5A).
Conversely, ['251]GF-II did not show an increase over binding obtained on
vitronectin coated wells alone, until concentrations of 11 ng/l00 1.1L - 33
ng/l00
tL were reached (FIG. 5B).
Preincubation of IGFBP-3 with either ['251]GF-I or [125I]-IGF-ll
did not alter the binding to vitronectin as compared to the non pre-incubated
concentrations of IGFBP-3 and [1251]GF-I or ['251]-IGF-II.
EXAMPLE 4
Binding of IGF-I to vitronectin in the presence of recombinant IGFBPs
produced in mammalian cells
Binding assays examining the ability of ['251]-IGF-I to bind to VN-coated
dishes in
the presence of IGFBPs, added at the same time as the radiolabel, were
performed
as described in Upton et a/.,1 999, supra. The data are shown in FIG. 6.
Increasing
amounts of IGFBP-2, -4 and -5 resulted in increased binding of labelled IGF-I
to
vitronectin coated wells. At the highest amount of IGFBP tested, 5 ng, binding
of
labelled IGF-I was increased approximately 2.8-, 3.8- and 8-fold for IGFBP-2,
4
and 5 compared to control wells where VN, but no IGFBPs, were present.
The presence of IGFBP-3 at 0.05, 0.2 and 0.5 ng/well also
increased binding of labelled IGF-I to VN while 2 and 5 ng of IGFBP-3/well
appeared to inhibit binding of the radiolabel. All concentrations of IGFBP-1
and -
6 tested were also inhibitory.

CA 02424394 2003-03-21
WO 02/24219 PCT/AU01/01196
29
These results demonstrate that unlike the situation with IGF-II,
minimal direct binding of IGF-I to VN is observed. However, the presence of
IGFBPs, especially IGFBP-2, -3, -4 and -5 enhances IGF binding to VN-coated
wells, suggesting that IGFBPs mediate the binding of IGF-I to VN in this
situation. In addition, the data suggests that IGFBPs have the potential to
both
enhance and inhibit binding of IGF-I to VN, depending on a) which IGFBPs are
present and b) the amount of IGFBP present.
EXAMPLE 5
Binding of labelled IGF-I to VN in the presence IGFBP-3 variants
Binding assays examining the ability of [1254-IGF-I to bind to VN-coated
dishes in
the presence of an IGFBP-3 variant in which the putative "heparin binding
domain" was mutated (IGFBP-3 HBD) and a variant in which glycosylation sites
had been mutated (Non-gly IGFBP-3) were performed as described above. The
data are shown in FIG. 7.
The IGFBP-3 glycosylation mutant used in these binding studies had three
potential 0-glycosylation sites at positions Asn89, Asn109, Asn172 mutated to
Ala.
The IGFBP-3 HBD mutant did not enhance binding of ['251]GF-I to VN-
coated dishes, suggesting the heparin binding domain of IGFBP-3 is involved in
binding IGFBP-3 to VN. Other studies have identified that amino acid residues
outside of the putative "heparin binding domain" are responsible for IGF-I
binding
to IGFBP-3, hence it is most likely that the results represent decreased
binding of
IGFBP-3 to VN, rather than decreased binding of labelled IGF-I to IGFBP-3
(Imai
et a, 2000, J Biol Chem 275:18188-18194).
Interestingly, the non-glycosylated IGFBP-3 mutant significantly
enhanced binding of ['251]-IGF-I to bind to VN-coated dishes. The striking 20-
fold
increase in binding of labelled IGF-I to VN-coated wells in the presence of 2
ng of
the mutant IGFBP-3 is intriguing and suggests that glycosylation of IGFBP-3
inhibits interaction of either a) IGF-I to IGFBP-3 or b) IGFBP-3 with VN.
Alternatively, both interactions may be hindered by the presence of
carbohydrates.
Whether non-glycoslyated IGFBP-3 is functionally relevant in vivo
remains to be established. Nevertheless, this finding suggests that non-

CA 02424394 2003-03-21
WO 02/24219 PCT/AU01/01196
glycosylated IGFBP-3 bound to VN may be a useful way to deliver IGF-I to sites
where IGF-I is required to potentiate cell function such as in stimulating
cell
proliferation. Alternatively, non-glycosylated IGFBP-3 bound to VN may provide
a mechanism for sequestering excess IGF-I in situations where cell
proliferation is
5 not required such as in tumours overexpressing IGFs.
EXAMPLE 6
Competition binding assay assessing ability of IGF variants to compete with
labelled
IGFs for binding to VN in the presence of non-glycosylated recombinant IGFBP-3
The concentration of IGFBP3 that produced the highest binding of radiolabelled
10 IGF depicted in Figure 5 was used in a competition assay to determine
whether
increasing concentrations of IGFs and desIGFs could compete with radiolabelled
IGFs for binding to vitronectin in the presence of IGFBP3. The results
indicated
that IGF-I, IGF-II, des(1-6)IGF-II and perhaps also des(1-3)IGF-I (not shown),
at
high concentrations, can compete with the binding of [1251]-IGF-11 to
vitronectin in
15 the presence of 10 ng of IGFBP3 (FIG. 8). These experimental results
indicated
that only IGF-I,-II and des(1-6) IGF-II could compete with for binding to
vitronectin coated wells in the presence of 30 ng of IGFBP3.
EXAMPLE 7
Stimulation of cell proliferation by isolated protein complexes comprising
20 IGF-II and vitronectin
The strategy of pre-binding IGF-II to VN was used in this study in an attempt
to
more accurately reflect the extracellular environment in vivo. Most cell
culture
approaches add exogenous substrates in solution phase; thereby the cells are
constantly exposed to the treatments. Cells in tissues do not encounter this
25 "constant, solution phase" environment in vivo. Thus the approach
adopted for this
study was to pre-bind IGFs to VN, a situation which more accurately mirrors
the
in vivo conditions.
Following pre-binding of IGFs to VN in culture dishes, cells were
seeded into wells and the, ability of IGFs complexed to VN to stimulate
protein
30 synthesis was examined by established methods (Francis et al., 1986,
Biochem J.
233:207-213). Increased protein synthesis correlates with increases in cell
number,
hence is a reflection of cell proliferation. Responses, expressed as
percentage

CA 02424394 2003-03-21
WO 02/24219 PCT/AU01/01196
31
above control wells in which no \TN or IGFs were present, measured the
incorporation of [314]4eucine into newly synthesised protein over 48 hrs.
Referring to the data in FIG. 9, when 3, 10, 30, 100, 300 and 1000
ng of IGF-11 was pre-bound to the wells in the absence of VN, resulting
responses
of 8, 12, 10, 16, 24 and 43% above the control wells (-VN, -IGF) respectively
were observed. Furthermore, these same doses of IGF-II pre-bound to VN coated
wells stimulated incorporation of [311]-1eucine into protein with effects of
19, 29,
39, 51, 70 and 101% respectively. Combining the responses obtained with VN
alone (12%) with that obtained for alone
(refer to above) gives rise to
predicted additive effects of 20, 24, 22, 28, 36 and 55% for 3, 10, 30, 100,
300 and
100 ng of IGF-II respectively. These values are significantly different
(p<0.05) to
the actual effects observed when IGF-II was pre-bound to VN at all doses
except
for the two lowest amounts of IGF-II tested (3 and 10 ng). Thus IGF-11 pre-
bound
to VN stimulates synergistic effects in protein synthesis ranging from 5 to
46%
greater than the calculated additive effects. These responses may well be a
result
of the direct binding of IGF-IE to VN, and may also arise from indirect
binding of
to VN via IGFBPs. HaCAT keratinocytes produce large amounts of
IGFBP-3 (Wraight et al., 1994, J. Invest. Dermatol 103:627-631).
EXAMPLE 8
Binding of labelled IGF-II to VN in the presence of recombinant IGFBPs
produced in mammalian cells
Binding assays examining the ability of ['251]GF-II to bind to VN-coated
dishes
in the presence of IGFBPs, added at the same time as the radiolabel, were
performed as described in Upton et al., 1999, supra. The IGFBPs used in these
studies were glycosylated having been produced in mammulian cells. As shown in
FIG. 10, increasing amounts of IGFBP-1, -3 and -6 resulted in decreased
binding
of labelled IGF-I1 to vitronectin coated wells in a dose-dependent manner.
IGFBP-
2 also appeared to compete for binding of labelled IGF-II to VN, albeit less
effectively than IGFBP-1, -3 or -6. IGFBP-4 on the other hand had little
effect on
binding of IGF-II to \TN, while IGFBP-5 appeared to enhance IGF-II binding to
a
small extent. The inhibitory effect of IGFBP-3 could result from IGFBP-3
competing for binding of IGF-II to the same binding region on VN.
Alternatively,

CA 02424394 2003-03-21
WO 02/24219 PCT/AU01/01196
32
or in addition, the inhibitory effect of IGFBP-3, as well as IGFBP-1 and -6.
may
arise from the affinity of IGF-I1 for VN being less than the affinity of IGF-
II for
these IGFBPs, hence these IGFBPs sequester IGF-I1 and the complex does not
bind to VN.
EXAMPLE 9
Stimulation of cell proliferation by isolated protein complexes comprising
IGF-I, IGFBP-5 and vitronectin
Referring to Table 2, IGF-I with IGFBP-5 and vitronectin stimulated
keratinocyte
proliferation (as measured by 3H-leucine incorporation into newly synthesized
protein) at all concentrations tested with synergistic effects observed at the
highest
amount tested.
It is proposed by the present inventors that the effect of isolated
protein complexes upon cell proliferation may be even greater at higher
concentrations of IGFBP-5 than the relatively low amount described in Table 2.
EXAMPLE 10
Engineering IGF and IGFBP variants
Amino acid residues in IGFBP-3 that are important for association with the ECM
and have been defined as the putative "heparin-binding domain" are residues
KGRKR at positions 228-232. Similar residues are found in the corresponding
region of IGFBP-5. The IGFBP-3 HBD mutant that was used in the binding
studies reported herein had the residues KGRKR altered to MDGEA based on the
amino acids found in the corresponding positions in IGFBP-1. These changes
result in a charge reversal in this part of the protein. This mutant still
binds IGF-I
and with high affinity but binds the acid-labile subunit and the
cell surface
poorly (Firth et al., 1998, J. Biol. Chem. 273 2631-2638).
Heparin binding motifs in a diverse range of proteins were
originally described in Cardin et al., 1989, Arteriosclerosis 9 21-32.
The C-domain of human IGF-II contains a number of positively
charged amino acid residues and in particular positions 34-40 contains the
amino
acids RVSRRSR. Given that positively charged amino acids are important in
mediating binding IGFBPs to cell surfaces and to VN, these amino acids in IGF-
I1
may be important in binding of IGF-II directly to VN. Regardless, it would be
a

CA 02424394 2003-03-21
WO 02/24219 PCT/AU01/01196
33
relatively simple procedure to introduce a "heparin binding motif' similar to
that
found in IGFBP-3 (BXBBB; where B is a basic amino acid) by creating an
mutant with deletions of either V' or 836 along with 839. The importance of
positive
residues in mediating binding of to VN
is further illustrated by the present
inventors' evidence of reduced binding of the chicken IGF-II mutant, (dese)-
IGF-11, to VN.
The C-domain of IGF-I on the other hand contains a relatively non-
charged stretch of amino acids in the corresponding region of the protein to
that
described above for IGF-II. This may explain why IGF-I does not bind directly
to
VN. In addition, insulin, which also does not bind to VN (or to IGFBPs) does
not
have a corresponding C-domain as it is cleaved out in the mature protein.
Human IGF-I SSSRRAPQT
Human IGF-11 RVSRRS- - R
Introduction of the IGF-II sequence RVSRRSR or the IGFBP3
sequence KGRKR into IGF-I could enable IGF-I to bind VN directly.
EXAMPLE 11
Isolated protein complexes, cell proliferation and survival
Bc1-2 transcription, a critical element of the cell survival pathway, is
elevated in
cells that attach to VN through alphav-beta3 integrins. (Matter & Ruoslahti,
2001,
J. Biol. Chem. 276 27757-27763). In addition, IGF-I protects cells from
apoptosis
by elevating bc1-2 transcription in an AKT-dependent manner (Pugazhenthi et
a/. ,1999,. J Biol. Chem. 274 27529-35). The IGF receptor physically
associates
with the alphav-beta3 integrin with a synergistic effect on cell growth
(Schneller
et a/.,1997, EMBO J 16 5600-5607). Thus isolated protein complexes of the
invention may provide an extracellular point of integration for initiating the
cell
survival signals mediated by both the integrin and the growth factor receptor.
IGFBP-5 has been demonstrated to potentiate the anti-apoptotic
and mitogenic effects of IGF-I in prostate cancer cells (Miyake et al., 2000,
Endocrinol. 141 2257-2265). In addition, the synthesis of VN in vivo by glioma
cells and in colorectal adeno carcinoma correlates with tumour grade (Uhm et
al.,
1999, Clin Cancer Res. 5 1587-1594; Tomasini-Johansson et al., 1994, Exp Cell
Res. 214 303-312; Gladson et al., 1995, J. Cell Sci. 108 947-56; Gladson &

CA 02424394 2003-03-21
WO 02/24219 PCT/AU01/01196
34
Cheresh, 1991, J. Clin. Invest. 88 1924-32).
Hence, according to the present invention, it is proposed that
IGF:IGFBP:VN complexes formed in vivo may promote tumour cell survival and
progression. The invention therefore contemplates therapeutic agents that
disrupt
in vivo complex formation.
The heparin-binding domain of VN has been reported to inhibit
fibronectin matrix assembly (Hocking et al., 1999, J. Biol. Chem. 274 27257-
27264). Reduced fibronectin deposition is associated with tumour cell invasion
as
decreased cell migration rates are associated with increased levels of
polymerised
fibronectin (Mona et al., 1994, Nature 367 193-196). Hence, IGF:IGFBP
complexes bound to the heparin binding domain of VN, may dampen fibronectin
matrix assembly and facilitate tumour invasion of local connective tissue.
Thus,
the invention contemplates therapeutic agents that disrupt these in vivo
complexes
to reduce tumour invasiveness.
Isolated protein complexes and wound healing
The converse argument can be used to support the use of the complex in
situations
where cell migration is required such as in wound repair. The IGF system plays
an
important role in wound healing and both IGF-I and IGFBP-3 are present in
wound fluid in significant concentrations. (Skottner et al., 1990, Acta Scand.
Suppl. 367 63-66; Clark R (ed) 1996, Molecular and Cell Biology of Wound
Repair, pp 3-50, Plenum Press, New York; Robertson et al., 1996, Endocrinol.
137 2774-2784; Vogt et a/.,1998, Growth Horm. IGF Res. 8 Suppl B:107-9.
IGFBPs have been demonstrated to reduce the rate of IGF
clearance from wounds. (Robertson et al., 1999,. Am J Physiol. 276 E663-71).
IGFBP-3:IGF-I complexes bind to fibrin clots in vitro leading to the
suggestion
this also occurs in vivo, resulting in concentration of IGF-I at wound sites.
(Campbell et al., 1999, J. Biol. Chem. 274 30215-30221). Similarly,
vitronectin
binds to fibrin (Podor et al., 2000, J. Biol. Chem. 275 19788-19794). It is
also
noted that vironectin-null mice exhibit increased wound fibrinolysis and
decreased
microvascular angiogenesis (Jang et al., 2000, Surgery 127 696-704).
According to the present invention, it is proposed that IGFs bound
to IGFBPs can bind to VN, which in turn associates with the fibrin clot, thus

CA 02424394 2003-03-21
WO 02/24219 PCT/AU01/01196
providing a reservoir of IGFs at the wound site. Thus isolated protein
complexes
of the invention could be administered to wounds to accelerate the repair
process.
A particular aspect of would healing contemplated by the present
invention relates to healing diabetic foot ulcers. Wound healing is delayed in
5 diabetes. Growth factors influence the healing process and in
particular, IGFs have
been shown to stimulate keratinocyte proliferation. However, analysis of
tissues
from diabetic skin and foot ulcers reveals lack of expression of IGF-I within
the
basal layer and fibroblasts compared to tissue sections from non-diabetic
patients.
(Blakytny et at., 2000, J. Pathol. 190 589-594). Isolated protein complexes of
the
10 invention could be useful in delivery of IGF-I to these types of wounds.
Isolated protein complexes and bone engineering
IGFBP-5 facilitates binding of labelled IGF-I to bone by a mechanism that is
independent of IGF receptors. (Mohan et cd.,1995,. J. Biol. Chem 270 20424-
20431) and enhances IGF-stimulated osteoblast function. (Andress, 1995, J.
Biol.
15 Chem. 270 28289-28296).
The implant material hydroxylapatite has been shown in numerous
studies to be highly biocomaptible and to osseointegrate well with existing
bone.
Recent evidence has found that hydroxylapatite will absorb more VN from serum
than other commonly used implant materials such as titanium and stainless
steel.
20 The absorption of VN was accompanied by greater binding of osteoblast
precursor
cells (Kilpadi et at., 2001, J. Biomed. Mater. Res. 57 258-267).
The effect of VN on nanophase alumina vs conventional alumina
was recently examined with VN being found to enhance osteoblast adhesion
(Webster et at., 2001, Tissue Eng 7 291-301).
25 The present inventors propose that isolated protein
complexes of
the invention may useful as coating applied to these materials and thereby
accelerate bone cell attachment, growth and integration in orthopaedic
applications such as hip joint replacements.
VN enhances IGF-I stimulated osteoclastic resorption and
30 proteinase activities in rabbit bone cell culture. (Rousselle et at.,
2001, Histology
& Histopathology 16 727-734) and IGFBP-5 enhances IGF-stimulated osteoblast
mitogenesis. (Andress & Birnbaum, 1992, J. Biol. Chem. 267 22467-22472).

CA 02424394 2010-05-31
36
The present inventors propose that as the major IGFBP produced by bone cells
is
IGFBP-5, the potentiation of the IGF effect by VN is likely to also involve
IGFBP-5.
Isolated protein complexes and treatment of atherosclerosis
IGF-I has previously been implicated in the development of experimental
atherosclerotic
lesions. Moreover, alphav-beta3 inhibitors have been demonstrated to reduce
atherosclerotic
lesions - this being associated with inhibition of IGF-I-mediated signaling.
(Nichols et al., 1999,
Circ. Res. 85 1040-1045).
Given that:
(i) IGFBP-5 and VN are synthesised and secreted by arterial smooth muscle
cells
and are present in blood vessel walls; and
(ii) IGF:IGFBP-5:VN complexes promote vascular smooth muscle cell
mitogenesis
and migration (Nam & Clemmons, 2000, Growth Horm. IGF Res. 10:A23);
it is proposed by the present inventors that IGF:IGFBP-5:VN complexes are
involved in
formation of atherosclerotic lesions. Thus therapeutic agents that disrupt
complex formation
may hold potential in treating atheroslerosis.
Throughout the specification the aim has been to describe the preferred
embodiments of
the invention without limiting the invention to any one embodiment or specific
collection of
features. It will therefore be appreciated by those of skill in the art that,
in light of the instant
disclosure, various modifications and changes can be made in the particular
embodiments
exemplified without departing from the scope of the present invention.

CA 02424394 2003-03-21
WO 02/24219
PCT/AU01/01196
37
TABLE 1
Nucleic acid Reference
IGF-I Jansen et al., 1983, Nature 306 609
IGF-11 Jansen et al., 1985, FEBS Lett 179 243
IGFBP-1 Brinkman et al., 1988, EMBO J. 7 2417
IGFBP-2 Binkert et al., 1989, EMBO J. 8 2497
IGFBP-3 Wood et al., 1988, Mol. Endocrinol. 2 1176
IGFBP-4 LaTour et al., 1990, Mol. Endocrinol. 4 1806
IGFBP-5 Kiefer et al., 1991, Biochem. Biophys. Res. Comm. 176
219
IGFBP-6 Shimasaki et al., 1991, Mol. Endocrinol. 5 938
ALS Leong et al., 1992, Mol. Endocrinol. 6 870
Vitronectin Suzuki et al., 1985, EMBO J. 4 2519
TABLE 2
Treatment - Vitronectin + Vitronectin
Control 100 2.6
IGFBP-5 109.5 9.2
100 ng IGF-1 + IGFBP-5 117.4 10.2 119.4 1.9
300 ng IGF-1 + IGFBP-5 140.9 2.0 154.3 1.8
1000 ng IGF-1 + IGFBP-5 144.3 11.0 161.4 9.1

Representative Drawing

Sorry, the representative drawing for patent document number 2424394 was not found.

Administrative Status

2024-08-01:As part of the Next Generation Patents (NGP) transition, the Canadian Patents Database (CPD) now contains a more detailed Event History, which replicates the Event Log of our new back-office solution.

Please note that "Inactive:" events refers to events no longer in use in our new back-office solution.

For a clearer understanding of the status of the application/patent presented on this page, the site Disclaimer , as well as the definitions for Patent , Event History , Maintenance Fee  and Payment History  should be consulted.

Event History

Description Date
Time Limit for Reversal Expired 2020-09-24
Change of Address or Method of Correspondence Request Received 2020-04-28
Common Representative Appointed 2019-10-30
Common Representative Appointed 2019-10-30
Letter Sent 2019-09-24
Letter Sent 2018-07-04
Inactive: Single transfer 2018-06-20
Maintenance Request Received 2017-08-25
Grant by Issuance 2017-05-23
Inactive: Cover page published 2017-05-22
Inactive: Office letter 2017-04-13
Notice of Allowance is Issued 2017-04-13
Inactive: Q2 passed 2017-04-03
Inactive: Approved for allowance (AFA) 2017-04-03
Letter Sent 2017-03-23
Reinstatement Request Received 2017-03-15
Pre-grant 2017-03-15
Withdraw from Allowance 2017-03-15
Final Fee Paid and Application Reinstated 2017-03-15
Inactive: Final fee received 2017-03-15
Maintenance Request Received 2016-08-25
Deemed Abandoned - Conditions for Grant Determined Not Compliant 2016-03-17
Notice of Allowance is Issued 2015-09-17
Letter Sent 2015-09-17
Notice of Allowance is Issued 2015-09-17
Maintenance Request Received 2015-08-28
Letter Sent 2015-08-24
Letter Sent 2015-08-24
Inactive: Multiple transfers 2015-08-12
Inactive: QS passed 2015-08-12
Inactive: Approved for allowance (AFA) 2015-08-12
Amendment Received - Voluntary Amendment 2014-12-24
Maintenance Request Received 2014-09-09
Inactive: S.30(2) Rules - Examiner requisition 2014-06-25
Inactive: Report - No QC 2014-06-16
Amendment Received - Voluntary Amendment 2013-12-23
Maintenance Request Received 2013-09-11
Inactive: S.30(2) Rules - Examiner requisition 2013-06-25
Amendment Received - Voluntary Amendment 2013-02-12
Inactive: S.30(2) Rules - Examiner requisition 2012-08-23
Amendment Received - Voluntary Amendment 2011-11-30
Inactive: S.30(2) Rules - Examiner requisition 2011-06-01
Amendment Received - Voluntary Amendment 2010-05-31
Inactive: S.30(2) Rules - Examiner requisition 2009-11-30
Letter Sent 2006-10-05
All Requirements for Examination Determined Compliant 2006-09-13
Request for Examination Requirements Determined Compliant 2006-09-13
Request for Examination Received 2006-09-13
Letter Sent 2004-01-16
Inactive: Single transfer 2003-12-12
Inactive: Courtesy letter - Evidence 2003-06-23
Inactive: Cover page published 2003-06-19
Inactive: First IPC assigned 2003-06-17
Inactive: Notice - National entry - No RFE 2003-06-17
Application Received - PCT 2003-05-02
National Entry Requirements Determined Compliant 2003-03-21
Application Published (Open to Public Inspection) 2002-03-28

Abandonment History

Abandonment Date Reason Reinstatement Date
2017-03-15
2016-03-17

Maintenance Fee

The last payment was received on 2016-08-25

Note : If the full payment has not been received on or before the date indicated, a further fee may be required which may be one of the following

  • the reinstatement fee;
  • the late payment fee; or
  • additional fee to reverse deemed expiry.

Patent fees are adjusted on the 1st of January every year. The amounts above are the current amounts if received by December 31 of the current year.
Please refer to the CIPO Patent Fees web page to see all current fee amounts.

Owners on Record

Note: Records showing the ownership history in alphabetical order.

Current Owners on Record
FACTOR THERAPEUTICS LIMITED
Past Owners on Record
JENNIFER ANN KRICKER
ZEE UPTON
Past Owners that do not appear in the "Owners on Record" listing will appear in other documentation within the application.
Documents

To view selected files, please enter reCAPTCHA code :



To view images, click a link in the Document Description column. To download the documents, select one or more checkboxes in the first column and then click the "Download Selected in PDF format (Zip Archive)" or the "Download Selected as Single PDF" button.

List of published and non-published patent-specific documents on the CPD .

If you have any difficulty accessing content, you can call the Client Service Centre at 1-866-997-1936 or send them an e-mail at CIPO Client Service Centre.


Document
Description 
Date
(yyyy-mm-dd) 
Number of pages   Size of Image (KB) 
Description 2003-03-20 38 2,038
Drawings 2003-03-20 14 524
Claims 2003-03-20 4 173
Abstract 2003-03-20 1 55
Description 2010-05-30 38 1,965
Claims 2010-05-30 3 144
Claims 2011-11-29 3 149
Claims 2013-02-11 4 164
Claims 2013-12-22 4 164
Claims 2014-12-23 4 167
Reminder of maintenance fee due 2003-06-16 1 106
Notice of National Entry 2003-06-16 1 189
Courtesy - Certificate of registration (related document(s)) 2004-01-15 1 107
Reminder - Request for Examination 2006-05-24 1 116
Acknowledgement of Request for Examination 2006-10-04 1 176
Commissioner's Notice - Application Found Allowable 2015-09-16 1 162
Courtesy - Abandonment Letter (NOA) 2016-04-27 1 163
Notice of Reinstatement 2017-03-22 1 169
Courtesy - Certificate of registration (related document(s)) 2018-07-03 1 125
Maintenance Fee Notice 2019-11-04 1 177
PCT 2003-03-20 22 1,034
Correspondence 2003-06-16 1 24
Fees 2003-09-09 1 34
Fees 2004-08-29 1 35
Fees 2005-08-16 1 34
Fees 2006-08-31 1 31
Fees 2007-08-19 1 34
Fees 2008-08-28 1 34
Fees 2009-09-17 1 36
Fees 2010-09-15 1 41
Fees 2011-09-11 1 39
Fees 2012-09-20 1 39
Fees 2013-09-10 1 41
Fees 2014-09-08 1 41
Maintenance fee payment 2015-08-27 1 42
Maintenance fee payment 2016-08-24 1 41
Reinstatement 2017-03-14 1 45
Final fee 2017-03-14 1 45
Courtesy - Office Letter 2017-04-12 1 45
Maintenance fee payment 2017-08-24 1 40